101
|
Affiliation(s)
- Seungbok Yang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Yoonjae Cho
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Jiwon Jang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
102
|
Roth JG, Huang MS, Li TL, Feig VR, Jiang Y, Cui B, Greely HT, Bao Z, Paşca SP, Heilshorn SC. Advancing models of neural development with biomaterials. Nat Rev Neurosci 2021; 22:593-615. [PMID: 34376834 PMCID: PMC8612873 DOI: 10.1038/s41583-021-00496-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells have emerged as a promising in vitro model system for studying the brain. Two-dimensional and three-dimensional cell culture paradigms have provided valuable insights into the pathogenesis of neuropsychiatric disorders, but they remain limited in their capacity to model certain features of human neural development. Specifically, current models do not efficiently incorporate extracellular matrix-derived biochemical and biophysical cues, facilitate multicellular spatio-temporal patterning, or achieve advanced functional maturation. Engineered biomaterials have the capacity to create increasingly biomimetic neural microenvironments, yet further refinement is needed before these approaches are widely implemented. This Review therefore highlights how continued progression and increased integration of engineered biomaterials may be well poised to address intractable challenges in recapitulating human neural development.
Collapse
Affiliation(s)
- Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Thomas L Li
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Vivian R Feig
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Yuanwen Jiang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Henry T Greely
- Stanford Law School, Stanford University, Stanford, CA, USA
| | - Zhenan Bao
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
103
|
Galgoczi S, Ruzo A, Markopoulos C, Yoney A, Phan-Everson T, Li S, Haremaki T, Metzger JJ, Etoc F, Brivanlou AH. Huntingtin CAG expansion impairs germ layer patterning in synthetic human 2D gastruloids through polarity defects. Development 2021; 148:272380. [PMID: 34608934 PMCID: PMC8513611 DOI: 10.1242/dev.199513] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG repeats in the huntingtin gene (HTT). Although HD has been shown to have a developmental component, how early during human embryogenesis the HTT-CAG expansion can cause embryonic defects remains unknown. Here, we demonstrate a specific and highly reproducible CAG length-dependent phenotypic signature in a synthetic model for human gastrulation derived from human embryonic stem cells (hESCs). Specifically, we observed a reduction in the extension of the ectodermal compartment that is associated with enhanced activin signaling. Surprisingly, rather than a cell-autonomous effect, tracking the dynamics of TGFβ signaling demonstrated that HTT-CAG expansion perturbs the spatial restriction of activin response. This is due to defects in the apicobasal polarization in the context of the polarized epithelium of the 2D gastruloid, leading to ectopic subcellular localization of TGFβ receptors. This work refines the earliest developmental window for the prodromal phase of HD to the first 2 weeks of human development, as modeled by our 2D gastruloids. Summary: 2D gastruloids of isogenic human embryonic stem cells modeling Huntington's Disease reveal that huntingtin CAG expansion perturbs the spatial restriction of the activin response in the context of the polarized epithelium.
Collapse
Affiliation(s)
- Szilvia Galgoczi
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Albert Ruzo
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Christian Markopoulos
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Yoney
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Tien Phan-Everson
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Shu Li
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Tomomi Haremaki
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Jakob J Metzger
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
104
|
Hoang P, Ma Z. Biomaterial-guided stem cell organoid engineering for modeling development and diseases. Acta Biomater 2021; 132:23-36. [PMID: 33486104 PMCID: PMC8629488 DOI: 10.1016/j.actbio.2021.01.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/30/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Organoids are miniature models of organs to recapitulate spatiotemporal cellular organization and tissue functionality. The production of organoids has revolutionized the field of developmental biology, providing the possibility to study and guide human development and diseases in a dish. More recently, novel biomaterial-based culture systems demonstrated the feasibility and versatility to engineer and produce the organoids in a consistent and reproducible manner. By engineering proper tissue microenvironment, functional organoids have been able to exhibit spatial-distinct tissue patterning and morphogenesis. This review focuses on enabling technologies in the field of organoid engineering, including the control of biochemical and biophysical cues via hydrogels, as well as size and geometry control via microwell and microfabrication techniques. In addition, this review discusses the enhancement of organoid systems for therapeutic applications using biofabrication and organoid-on-chip platforms, which facilitate the assembly of complex organoid systems for in vitro modeling of development and diseases. STATEMENT OF SIGNIFICANCE: Stem cell organoids have revolutionized the fields of developmental biology and tissue engineering, providing the opportunity to study human organ development and disease progression in vitro. Various works have demonstrated that organoids can be generated using a wide variety of engineering tools, materials, and systems. Specific culture microenvironment is tailored to support the formation, function, and physiology of the organ of interest. This review highlights the importance of cellular microenvironment in organoid culture, the versatility of organoid engineering techniques, and future perspectives to build better organoid systems.
Collapse
Affiliation(s)
- Plansky Hoang
- Department of Biomedical and Chemical Engineering, Syracuse University, NY, United States; BioInspired Syracuse Institute for Material and Living Systems, NY, United States
| | - Zhen Ma
- Department of Biomedical and Chemical Engineering, Syracuse University, NY, United States; BioInspired Syracuse Institute for Material and Living Systems, NY, United States.
| |
Collapse
|
105
|
Steventon B, Busby L, Arias AM. Establishment of the vertebrate body plan: Rethinking gastrulation through stem cell models of early embryogenesis. Dev Cell 2021; 56:2405-2418. [PMID: 34520764 DOI: 10.1016/j.devcel.2021.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/20/2021] [Accepted: 08/14/2021] [Indexed: 12/28/2022]
Abstract
A striking property of vertebrate embryos is the emergence of a conserved body plan across a wide range of organisms through the process of gastrulation. As the body plan unfolds, gene regulatory networks (GRNs) and multicellular interactions (cell regulatory networks, CRNs) combine to generate a conserved set of morphogenetic events that lead to the phylotypic stage. Interrogation of these multilevel interactions requires manipulation of the mechanical environment, which is difficult in vivo. We review recent studies of stem cell models of early embryogenesis from different species showing that, independent of species origin, cells in culture form similar structures. The main difference between embryos and in vitro models is the boundary conditions of the multicellular ensembles. We discuss these observations and suggest that the mechanical and geometric boundary conditions of different embryos before gastrulation hide a morphogenetic ground state that is revealed in the stem-cell-based models of embryo development.
Collapse
Affiliation(s)
| | - Lara Busby
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Alfonso Martinez Arias
- Systems Bioengineering, DCEXS, Universidad Pompeu Fabra, Doctor Aiguader, 88 ICREA, Pag Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
106
|
Miloradovic D, Pavlovic D, Jankovic MG, Nikolic S, Papic M, Milivojevic N, Stojkovic M, Ljujic B. Human Embryos, Induced Pluripotent Stem Cells, and Organoids: Models to Assess the Effects of Environmental Plastic Pollution. Front Cell Dev Biol 2021; 9:709183. [PMID: 34540831 PMCID: PMC8446652 DOI: 10.3389/fcell.2021.709183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 02/03/2023] Open
Abstract
For a long time, animal models were used to mimic human biology and diseases. However, animal models are not an ideal solution due to numerous interspecies differences between humans and animals. New technologies, such as human-induced pluripotent stem cells and three-dimensional (3D) cultures such as organoids, represent promising solutions for replacing, refining, and reducing animal models. The capacity of organoids to differentiate, self-organize, and form specific, complex, biologically suitable structures makes them excellent in vitro models of development and disease pathogenesis, as well as drug-screening platforms. Despite significant potential health advantages, further studies and considerable nuances are necessary before their clinical use. This article summarizes the definition of embryoids, gastruloids, and organoids and clarifies their appliance as models for early development, diseases, environmental pollution, drug screening, and bioinformatics.
Collapse
Affiliation(s)
- Dragana Miloradovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Sandra Nikolic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Milos Papic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Milivojevic
- Laboratory for Bioengineering, Department of Science, Institute for Information Technologies, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag Stojkovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- SPEBO Medical Fertility Hospital, Leskovac, Serbia
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
107
|
Girgin MU, Broguiere N, Hoehnel S, Brandenberg N, Mercier B, Arias AM, Lutolf MP. Bioengineered embryoids mimic post-implantation development in vitro. Nat Commun 2021; 12:5140. [PMID: 34446708 PMCID: PMC8390504 DOI: 10.1038/s41467-021-25237-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The difficulty of studying post-implantation development in mammals has sparked a flurry of activity to develop in vitro models, termed embryoids, based on self-organizing pluripotent stem cells. Previous approaches to derive embryoids either lack the physiological morphology and signaling interactions, or are unconducive to model post-gastrulation development. Here, we report a bioengineering-inspired approach aimed at addressing this gap. We employ a high-throughput cell aggregation approach to simultaneously coax mouse embryonic stem cells into hundreds of uniform epiblast-like aggregates in a solid matrix-free manner. When co-cultured with mouse trophoblast stem cell aggregates, the resulting hybrid structures initiate gastrulation-like events and undergo axial morphogenesis to yield structures, termed EpiTS embryoids, with a pronounced anterior development, including brain-like regions. We identify the presence of an epithelium in EPI aggregates as the major determinant for the axial morphogenesis and anterior development seen in EpiTS embryoids. Our results demonstrate the potential of EpiTS embryoids to study peri-gastrulation development in vitro.
Collapse
Affiliation(s)
- Mehmet U Girgin
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Biozentrum, University of Basel, 4056, Basel, Switzerland
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sylke Hoehnel
- SUN bioscience, EPFL Innovation Park, Lausanne, Switzerland
| | | | - Bastien Mercier
- Faculty of Medicine and Pharmacy, University of Grenoble Alpes, Grenoble, France
| | | | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Roche Institute for Translational Bioengineering (ITB), Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland.
| |
Collapse
|
108
|
From Bipotent Neuromesodermal Progenitors to Neural-Mesodermal Interactions during Embryonic Development. Int J Mol Sci 2021; 22:ijms22179141. [PMID: 34502050 PMCID: PMC8431582 DOI: 10.3390/ijms22179141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
To ensure the formation of a properly patterned embryo, multiple processes must operate harmoniously at sequential phases of development. This is implemented by mutual interactions between cells and tissues that together regulate the segregation and specification of cells, their growth and morphogenesis. The formation of the spinal cord and paraxial mesoderm derivatives exquisitely illustrate these processes. Following early gastrulation, while the vertebrate body elongates, a population of bipotent neuromesodermal progenitors resident in the posterior region of the embryo generate both neural and mesodermal lineages. At later stages, the somitic mesoderm regulates aspects of neural patterning and differentiation of both central and peripheral neural progenitors. Reciprocally, neural precursors influence the paraxial mesoderm to regulate somite-derived myogenesis and additional processes by distinct mechanisms. Central to this crosstalk is the activity of the axial notochord, which, via sonic hedgehog signaling, plays pivotal roles in neural, skeletal muscle and cartilage ontogeny. Here, we discuss the cellular and molecular basis underlying this complex developmental plan, with a focus on the logic of sonic hedgehog activities in the coordination of the neural-mesodermal axis.
Collapse
|
109
|
Mackinlay KML, Weatherbee BAT, Souza Rosa V, Handford CE, Hudson G, Coorens T, Pereira LV, Behjati S, Vallier L, Shahbazi MN, Zernicka-Goetz M. An in vitro stem cell model of human epiblast and yolk sac interaction. eLife 2021; 10:e63930. [PMID: 34403333 PMCID: PMC8370770 DOI: 10.7554/elife.63930] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Human embryogenesis entails complex signalling interactions between embryonic and extra-embryonic cells. However, how extra-embryonic cells direct morphogenesis within the human embryo remains largely unknown due to a lack of relevant stem cell models. Here, we have established conditions to differentiate human pluripotent stem cells (hPSCs) into yolk sac-like cells (YSLCs) that resemble the post-implantation human hypoblast molecularly and functionally. YSLCs induce the expression of pluripotency and anterior ectoderm markers in human embryonic stem cells (hESCs) at the expense of mesoderm and endoderm markers. This activity is mediated by the release of BMP and WNT signalling pathway inhibitors, and, therefore, resembles the functioning of the anterior visceral endoderm signalling centre of the mouse embryo, which establishes the anterior-posterior axis. Our results implicate the yolk sac in epiblast cell fate specification in the human embryo and propose YSLCs as a tool for studying post-implantation human embryo development in vitro.
Collapse
Affiliation(s)
- Kirsty ML Mackinlay
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
| | - Bailey AT Weatherbee
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
| | - Viviane Souza Rosa
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São PauloSão PauloBrazil
- MRC Laboratory of Molecular Biology, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Charlotte E Handford
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
- Centre for Trophoblast Research, University of CambridgeCambridgeUnited Kingdom
| | - George Hudson
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
| | - Tim Coorens
- Wellcome Sanger InstituteCambridgeUnited Kingdom
| | - Lygia V Pereira
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São PauloSão PauloBrazil
| | - Sam Behjati
- Wellcome Sanger InstituteCambridgeUnited Kingdom
| | - Ludovic Vallier
- Wellcome – MRC Cambridge Stem Cell Institute, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Marta N Shahbazi
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
- MRC Laboratory of Molecular Biology, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and NeuroscienceCambridgeUnited Kingdom
- Synthetic Mouse and Human Embryology Group, California Institute of Technology (Caltech), Division of Biology and Biological EngineeringPasadenaUnited States
| |
Collapse
|
110
|
Abstract
Micropatterning encompasses a set of methods aimed at precisely controlling the spatial distribution of molecules onto the surface of materials. Biologists have borrowed the idea and adapted these methods, originally developed for electronics, to impose physical constraints on biological systems with the aim of addressing fundamental questions across biological scales from molecules to multicellular systems. Here, I approach this topic from a developmental biologist's perspective focusing specifically on how and why micropatterning has gained in popularity within the developmental biology community in recent years. Overall, this Primer provides a concise overview of how micropatterns are used to study developmental processes and emphasises how micropatterns are a useful addition to the developmental biologist's toolbox.
Collapse
Affiliation(s)
- Guillaume Blin
- Institute for Regeneration and Repair, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
111
|
Brivanlou AH, Gleicher N. The evolution of our understanding of human development over the last 10 years. Nat Commun 2021; 12:4615. [PMID: 34326329 PMCID: PMC8322423 DOI: 10.1038/s41467-021-24793-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/29/2021] [Indexed: 12/30/2022] Open
Abstract
As it fulfills an irresistible need to understand our own origins, research on human development occupies a unique niche in scientific and medical research. In this Comment, we explore the progress in our understanding of human development over the past 10 years. The focus is on basic research, clinical applications, and ethical considerations.
Collapse
Affiliation(s)
- Ali H Brivanlou
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY, USA.
| | - Norbert Gleicher
- The Center for Human Reproduction, New York, NY, USA
- The Foundation for Reproductive Medicine, New York, NY, USA
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
112
|
van den Brink SC, van Oudenaarden A. 3D gastruloids: a novel frontier in stem cell-based in vitro modeling of mammalian gastrulation. Trends Cell Biol 2021; 31:747-759. [PMID: 34304959 DOI: 10.1016/j.tcb.2021.06.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022]
Abstract
3D gastruloids, aggregates of embryonic stem cells that recapitulate key aspects of gastrula-stage embryos, have emerged as a powerful tool to study the early stages of mammalian post-implantation development in vitro. Owing to their tractable nature and the relative ease by which they can be generated in large numbers, 3D gastruloids provide an unparalleled opportunity to study normal and pathological embryogenesis from a bottom-up perspective and in a high-throughput manner. Here, we review how gastruloid models can be exploited to deepen our understanding of mammalian development. In addition, we discuss current limitations, potential clinical applications, and ethical implications of this emerging model system.
Collapse
Affiliation(s)
- Susanne C van den Brink
- Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Alexander van Oudenaarden
- Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
113
|
Montanari MP, Tran NV, Shimmi O. Regulation of spatial distribution of BMP ligands for pattern formation. Dev Dyn 2021; 251:198-212. [PMID: 34241935 DOI: 10.1002/dvdy.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 12/25/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-ß (TGF-ß) family, have been shown to contribute to embryogenesis and organogenesis during animal development. Relevant studies provide support for the following concepts: (a) BMP signals are evolutionarily highly conserved as a genetic toolkit; (b) spatiotemporal distributions of BMP signals are precisely controlled at the post-translational level; and (c) the BMP signaling network has been co-opted to adapt to diversified animal development. These concepts originated from the historical findings of the Spemann-Mangold organizer and the subsequent studies about how this organizer functions at the molecular level. In this Commentary, we focus on two topics. First, we review how the BMP morphogen gradient is formed to sustain larval wing imaginal disc and early embryo growth and patterning in Drosophila. Second, we discuss how BMP signal is tightly controlled in a context-dependent manner, and how the signal and tissue dynamics are coupled to facilitate complex tissue structure formation. Finally, we argue how these concepts might be developed in the future for further understanding the significance of BMP signaling in animal development.
Collapse
Affiliation(s)
| | - Ngan Vi Tran
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|
114
|
Abstract
Morphogenesis is one of the most remarkable examples of biological pattern formation. Despite substantial progress in the field, we still do not understand the organizational principles responsible for the robust convergence of the morphogenesis process across scales to form viable organisms under variable conditions. Achieving large-scale coordination requires feedback between mechanical and biochemical processes, spanning all levels of organization and relating the emerging patterns with the mechanisms driving their formation. In this review, we highlight the role of mechanics in the patterning process, emphasizing the active and synergistic manner in which mechanical processes participate in developmental patterning rather than merely following a program set by biochemical signals. We discuss the value of applying a coarse-grained approach toward understanding this complex interplay, which considers the large-scale dynamics and feedback as well as complementing the reductionist approach focused on molecular detail. A central challenge in this approach is identifying relevant coarse-grained variables and developing effective theories that can serve as a basis for an integrated framework for understanding this remarkable pattern-formation process. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Yonit Maroudas-Sacks
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel;
| | - Kinneret Keren
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel; .,Network Biology Research Laboratories and The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
115
|
Kostopoulou N, Bellou S, Bagli E, Markou M, Kostaras E, Hyvönen M, Kalaidzidis Y, Papadopoulos A, Chalmantzi V, Kyrkou A, Panopoulou E, Fotsis T, Murphy C. Embryonic stem cells are devoid of macropinocytosis, a trafficking pathway for activin A in differentiated cells. J Cell Sci 2021; 134:jcs246892. [PMID: 34313314 DOI: 10.1242/jcs.246892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Ligand-receptor complexes formed at the plasma membrane are internalised via various endocytic pathways that influence the ultimate signalling output by regulating the selection of interaction partners by the complex along the trafficking route. We report that, in differentiated cells, activin A-receptor complexes are internalised via clathrin-mediated endocytosis (CME) and macropinocytosis (MP), whereas in human embryonic stem cells (hESCs) internalisation occurs via CME. We further show that hESCs are devoid of MP, which becomes functional upon differentiation towards endothelial cells through mesoderm mediators. Our results reveal, for the first time, that MP is an internalisation route for activin A in differentiated cells, and that MP is not active in hESCs and is induced as cells differentiate.
Collapse
Affiliation(s)
- Nikoleta Kostopoulou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Sofia Bellou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Confocal Laser Scanning Microscopy Unit, Network of Research Supporting Laboratories, University of Ioannina, Ioannina, 45110, Greece
| | - Eleni Bagli
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Maria Markou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Eleftherios Kostaras
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Yiannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Angelos Papadopoulos
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Varvara Chalmantzi
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Athena Kyrkou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Ekaterini Panopoulou
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Theodore Fotsis
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Carol Murphy
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham, A118 Aston Webb, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
116
|
Forsyth JE, Al-Anbaki AH, de la Fuente R, Modare N, Perez-Cortes D, Rivera I, Seaton Kelly R, Cotter S, Plusa B. IVEN: A quantitative tool to describe 3D cell position and neighbourhood reveals architectural changes in FGF4-treated preimplantation embryos. PLoS Biol 2021; 19:e3001345. [PMID: 34310594 PMCID: PMC8341705 DOI: 10.1371/journal.pbio.3001345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/05/2021] [Accepted: 07/01/2021] [Indexed: 11/30/2022] Open
Abstract
Architectural changes at the cellular and organism level are integral and necessary to successful development and growth. During mammalian preimplantation development, cells reduce in size and the architecture of the embryo changes significantly. Such changes must be coordinated correctly to ensure continued development of the embryo and, ultimately, a successful pregnancy. However, the nature of such transformations is poorly defined during mammalian preimplantation development. In order to quantitatively describe changes in cell environment and organism architecture, we designed Internal Versus External Neighbourhood (IVEN). IVEN is a user-interactive, open-source pipeline that classifies cells into different populations based on their position and quantifies the number of neighbours of every cell within a dataset in a 3D environment. Through IVEN-driven analyses, we show how transformations in cell environment, defined here as changes in cell neighbourhood, are related to changes in embryo geometry and major developmental events during preimplantation mammalian development. Moreover, we demonstrate that modulation of the FGF pathway alters spatial relations of inner cells and neighbourhood distributions, leading to overall changes in embryo architecture. In conjunction with IVEN-driven analyses, we uncover differences in the dynamic of cell size changes over the preimplantation period and determine that cells within the mammalian embryo initiate growth phase only at the time of implantation.
Collapse
Affiliation(s)
- Jessica E. Forsyth
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
- School of Mathematics, Alan Turing Building, University of Manchester, Manchester, United Kingdom
| | - Ali H. Al-Anbaki
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Roberto de la Fuente
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, Poland
| | - Nikkinder Modare
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Diego Perez-Cortes
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Isabel Rivera
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Rowena Seaton Kelly
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Simon Cotter
- School of Mathematics, Alan Turing Building, University of Manchester, Manchester, United Kingdom
| | - Berenika Plusa
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
117
|
Glykofrydis F, Cachat E, Berzanskyte I, Dzierzak E, Davies JA. Bioengineering Self-Organizing Signaling Centers to Control Embryoid Body Pattern Elaboration. ACS Synth Biol 2021; 10:1465-1480. [PMID: 34019395 DOI: 10.1021/acssynbio.1c00060] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Multicellular systems possess an intrinsic capacity to autonomously generate nonrandom state distributions or morphologies in a process termed self-organization. Facets of self-organization, such as pattern formation, pattern elaboration, and symmetry breaking, are frequently observed in developing embryos. Artificial stem cell-derived structures including embryoid bodies (EBs), gastruloids, and organoids also demonstrate self-organization, but with a limited capacity compared to their in vivo developmental counterparts. There is a pressing need for better tools to allow user-defined control over self-organization in these stem cell-derived structures. Here, we employ synthetic biology to establish an efficient platform for the generation of self-organizing coaggregates, in which HEK-293 cells overexpressing P-cadherin (Cdh3) spontaneously form cell clusters attached mostly to one or two locations on the exterior of EBs. These Cdh3-expressing HEK cells, when further engineered to produce functional mouse WNT3A, evoke polarized and gradual Wnt/β-catenin pathway activation in EBs during coaggregation cultures. The localized WNT3A provision induces nascent mesoderm specification within regions of the EB close to the Cdh3-Wnt3a-expressing HEK source, resulting in pattern elaboration and symmetry breaking within EBs. This synthetic biology-based approach puts us closer toward engineering synthetic organizers to improve the realism in stem cell-derived structures.
Collapse
Affiliation(s)
- Fokion Glykofrydis
- UK Centre for Mammalian Synthetic Biology, Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Elise Cachat
- UK Centre for Mammalian Synthetic Biology, Institute of Quantitative Biology, Biochemistry, and Biotechnology, The University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Ieva Berzanskyte
- UK Centre for Mammalian Synthetic Biology, Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Elaine Dzierzak
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Jamie A. Davies
- UK Centre for Mammalian Synthetic Biology, Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
118
|
Borsoi J, Farinha-Arcieri LE, Morato-Marques M, Delgado Sarafian R, Pinheiro M, Veiga Pereira L. Generation of genetically modified human induced pluripotent stem cell lines harboring haploin sufficient or dominant negative variants in the FBN1 gene. Stem Cell Res 2021; 54:102434. [PMID: 34174776 DOI: 10.1016/j.scr.2021.102434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/22/2021] [Accepted: 06/14/2021] [Indexed: 11/26/2022] Open
Abstract
Marfan Syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutations in the FBN1 gene. To investigate the molecular mechanisms of pathogenesis for the syndrome, we genetically modified the FBN1 gene in a line of induced pluripotent stem cells (hiPSCs) derived from a healthy donor using the CRISPR/Cas9 gene editing technology. The sublines described here were characterized according to established criteria and were shown to maintain pluripotency, three germ layer differentiation potential and genomic integrity. These clones can now be used to better understand the pathogenesis of MFS in different cell types.
Collapse
Affiliation(s)
- Juliana Borsoi
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil
| | - Luis Ernesto Farinha-Arcieri
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil
| | - Mariana Morato-Marques
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil
| | - Raquel Delgado Sarafian
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil
| | - Mara Pinheiro
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil
| | - Lygia Veiga Pereira
- National Laboratory for Embryonic Stem Cells (LaNCE), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP 05508-090, Brazil.
| |
Collapse
|
119
|
Zhu Y, Sazer D, Miller JS, Warmflash A. Rapid fabrication of hydrogel micropatterns by projection stereolithography for studying self-organized developmental patterning. PLoS One 2021; 16:e0245634. [PMID: 34077425 PMCID: PMC8172057 DOI: 10.1371/journal.pone.0245634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/29/2021] [Indexed: 11/30/2022] Open
Abstract
Self-organized patterning of mammalian embryonic stem cells on micropatterned surfaces has previously been established as an in vitro platform for early mammalian developmental studies, complimentary to in vivo studies. Traditional micropatterning methods, such as micro-contact printing (μCP), involve relatively complicated fabrication procedures, which restricts widespread adoption by biologists. Here, we demonstrate a rapid method of micropatterning by printing hydrogel micro-features onto a glass-bottomed culture vessel. The micro-features are printed using a projection stereolithography bioprinter yielding hydrogel structures that geometrically restrict the attachment of cells or proteins. Compared to traditional and physical photomasks, a digitally tunable virtual photomask is used in the projector to generate blue light patterns that enable rapid iteration with minimal cost and effort. We show that a protocol that makes use of this method together with LN521 coating, an extracellular matrix coating, creates a surface suitable for human embryonic stem cell (hESC) attachment and growth with minimal non-specific adhesion. We further demonstrate that self-patterning of hESCs following previously published gastrulation and ectodermal induction protocols achieves results comparable with those obtained with commercially available plates.
Collapse
Affiliation(s)
- Ye Zhu
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Daniel Sazer
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Jordan S. Miller
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail: (JSM); (AW)
| | - Aryeh Warmflash
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- Department of Biosciences, Rice University, Houston, Texas, United States of America
- * E-mail: (JSM); (AW)
| |
Collapse
|
120
|
Weatherbee BAT, Cui T, Zernicka-Goetz M. Modeling human embryo development with embryonic and extra-embryonic stem cells. Dev Biol 2021; 474:91-99. [PMID: 33333069 PMCID: PMC8232073 DOI: 10.1016/j.ydbio.2020.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022]
Abstract
Early human post-implantation development involves extensive growth combined with a series of complex morphogenetic events. The lack of precise spatial and temporal control over these processes leads to pregnancy loss. Given the ethical and technical limitations in studying the natural human embryo, alternative approaches are needed to investigate mechanisms underlying this critical stage of human development. Here, we present an overview of the different stem cells and stem cell-derived models which serve as useful, albeit imperfect, tools in understanding human embryogenesis. Current models include stem cells that represent each of the three earliest lineages: human embryonic stem cells corresponding to the epiblast, hypoblast-like stem cells and trophoblast stem cells. We also review the use of human embryonic stem cells to model complex aspects of epiblast morphogenesis and differentiation. Additionally, we propose that the combination of both embryonic and extra-embryonic stem cells to form three-dimensional embryo models will provide valuable insights into cell-cell chemical and mechanical interactions that are essential for natural embryogenesis.
Collapse
Affiliation(s)
- Bailey A T Weatherbee
- Mouse and Human Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and Neuroscience, Downing Street, Cambridge, CB2 3EG, UK
| | - Tongtong Cui
- Plasticity and Synthetic Embryology Group, California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA
| | - Magdalena Zernicka-Goetz
- Mouse and Human Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and Neuroscience, Downing Street, Cambridge, CB2 3EG, UK; Plasticity and Synthetic Embryology Group, California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA.
| |
Collapse
|
121
|
Affiliation(s)
- Jake Cornwall-Scoones
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA
| | - Magdalena Zernicka-Goetz
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA; Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and Neuroscience, Cambridge, CB2 3EG, UK.
| |
Collapse
|
122
|
Sozen B, Cornwall-Scoones J, Zernicka-Goetz M. The dynamics of morphogenesis in stem cell-based embryology: Novel insights for symmetry breaking. Dev Biol 2021; 474:82-90. [PMID: 33333067 PMCID: PMC8259461 DOI: 10.1016/j.ydbio.2020.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Breaking embryonic symmetry is an essential prerequisite to shape the initially symmetric embryo into a highly organized body plan that serves as the blueprint of the adult organism. This critical process is driven by morphogen signaling gradients that instruct anteroposterior axis specification. Despite its fundamental importance, what triggers symmetry breaking and how the signaling gradients are established in time and space in the mammalian embryo remain largely unknown. Stem cell-based in vitro models of embryogenesis offer an unprecedented opportunity to quantitatively dissect the multiple physical and molecular processes that shape the mammalian embryo. Here we review biochemical mechanisms governing early mammalian patterning in vivo and highlight recent advances to recreate this in vitro using stem cells. We discuss how the novel insights from these model systems extend previously proposed concepts to illuminate the extent to which embryonic cells have the intrinsic capability to generate specific, reproducible patterns during embryogenesis.
Collapse
Affiliation(s)
- Berna Sozen
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA; Yale University School of Medicine, Department of Genetics, New Haven, CT, 06510, USA.
| | - Jake Cornwall-Scoones
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA; Developmental Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Magdalena Zernicka-Goetz
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 E. California Boulevard, Pasadena, CA, 91125, USA; Mammalian Embryo and Stem Cell Group, University of Cambridge, Department of Physiology, Development and Neuroscience, Cambridge, CB2 3EG, UK.
| |
Collapse
|
123
|
Rosado-Olivieri EA, Brivanlou AH. Synthetic by design: Exploiting tissue self-organization to explore early human embryology. Dev Biol 2021; 474:16-21. [DOI: 10.1016/j.ydbio.2021.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
|
124
|
Gritti N, Oriola D, Trivedi V. Rethinking embryology in vitro: A synergy between engineering, data science and theory. Dev Biol 2021; 474:48-61. [DOI: 10.1016/j.ydbio.2020.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
|
125
|
Martyn I, Gartner ZJ. Expanding the boundaries of synthetic development. Dev Biol 2021; 474:62-70. [PMID: 33587913 PMCID: PMC8052276 DOI: 10.1016/j.ydbio.2021.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022]
Abstract
Embryonic tissue boundaries are critical to not only cement newly patterned structures during development, but also to serve as organizing centers for subsequent rounds of morphogenesis. Although this latter role is especially difficult to study in vivo, synthetic embryology offers a new vantage point and fresh opportunities. In this review, we cover recent progress towards understanding and controlling in vitro boundaries and how they impact synthetic model systems. A key point this survey highlights is that the outcome of self-organization is strongly dependent on the boundary imposed, and new insight into the complex functions of embryonic boundaries will be necessary to create better self-organizing tissues for basic science, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Iain Martyn
- Department of Pharmaceutical Chemistry, NSF Center for Cellular Construction, San Francisco, CA, USA
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, Chan Zuckerberg Biohub, NSF Center for Cellular Construction, San Francisco, CA, USA.
| |
Collapse
|
126
|
Reassembling gastrulation. Dev Biol 2021; 474:71-81. [DOI: 10.1016/j.ydbio.2020.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
|
127
|
Pereira Daoud AM, Popovic M, Dondorp WJ, Trani Bustos M, Bredenoord AL, Chuva de Sousa Lopes SM, van den Brink SC, Roelen BAJ, de Wert GMWR, Heindryckx B. Modelling human embryogenesis: embryo-like structures spark ethical and policy debate. Hum Reprod Update 2021; 26:779-798. [PMID: 32712668 DOI: 10.1093/humupd/dmaa027] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/06/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Studying the human peri-implantation period remains hindered by the limited accessibility of the in vivo environment and scarcity of research material. As such, continuing efforts have been directed towards developing embryo-like structures (ELS) from pluripotent stem cells (PSCs) that recapitulate aspects of embryogenesis in vitro. While the creation of such models offers immense potential for studying fundamental processes in both pre- and early post-implantation development, it also proves ethically contentious due to wide-ranging views on the moral and legal reverence due to human embryos. Lack of clarity on how to qualify and regulate research with ELS thus presents a challenge in that it may either limit this new field of research without valid grounds or allow it to develop without policies that reflect justified ethical concerns. OBJECTIVE AND RATIONALE The aim of this article is to provide a comprehensive overview of the existing scientific approaches to generate ELS from mouse and human PSCs, as well as discuss future strategies towards innovation in the context of human development. Concurrently, we aim to set the agenda for the ethical and policy issues surrounding research on human ELS. SEARCH METHODS The PubMed database was used to search peer-reviewed articles and reviews using the following terms: 'stem cells', 'pluripotency', 'implantation', 'preimplantation', 'post-implantation', 'blastocyst', 'embryoid bodies', 'synthetic embryos', 'embryo models', 'self-assembly', 'human embryo-like structures', 'artificial embryos' in combination with other keywords related to the subject area. The PubMed and Web of Science databases were also used to systematically search publications on the ethics of ELS and human embryo research by using the aforementioned keywords in combination with 'ethics', 'law', 'regulation' and equivalent terms. All relevant publications until December 2019 were critically evaluated and discussed. OUTCOMES In vitro systems provide a promising way forward for uncovering early human development. Current platforms utilize PSCs in both two- and three-dimensional settings to mimic various early developmental stages, including epiblast, trophoblast and amniotic cavity formation, in addition to axis development and gastrulation. Nevertheless, much hinges on the term 'embryo-like'. Extension of traditional embryo frameworks to research with ELS reveals that (i) current embryo definitions require reconsideration, (ii) cellular convertibility challenges the attribution of moral standing on the basis of 'active potentiality' and (iii) meaningful application of embryo protective directives will require rethinking of the 14-day culture limit and moral weight attributed to (non-)viability. Many conceptual and normative (dis)similarities between ELS and embryos thus remain to be thoroughly elucidated. WIDER IMPLICATIONS Modelling embryogenesis holds vast potential for both human developmental biology and understanding various etiologies associated with infertility. To date, ELS have been shown to recapitulate several aspects of peri-implantation development, but critically, cannot develop into a fetus. Yet, concurrent to scientific innovation, considering the extent to which the use of ELS may raise moral concerns typical of human embryo research remains paramount. This will be crucial for harnessing the potential of ELS as a valuable research tool, whilst remaining within a robust moral and legal framework of professionally acceptable practices.
Collapse
Affiliation(s)
- Ana M Pereira Daoud
- Department of Health Ethics and Society, Maastricht University, Maastricht, The Netherlands.,Department of Medical Humanities, Utrecht University Medical Center, Utrecht, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Mina Popovic
- Ghent-Fertility And Stem cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Wybo J Dondorp
- Department of Health Ethics and Society, Maastricht University, Maastricht, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.,School for Care and Public Health Research (CAPHRI), Maastricht University, Maastricht, The Netherlands.,Socrates chair Ethics of Reproductive Genetics endowed by the Dutch Humanist Association, Amsterdam, The Netherlands
| | - Marc Trani Bustos
- Ghent-Fertility And Stem cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium.,Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annelien L Bredenoord
- Department of Medical Humanities, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Susana M Chuva de Sousa Lopes
- Ghent-Fertility And Stem cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Susanne C van den Brink
- Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bernard A J Roelen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Guido M W R de Wert
- Department of Health Ethics and Society, Maastricht University, Maastricht, The Netherlands.,School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.,School for Care and Public Health Research (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Björn Heindryckx
- Ghent-Fertility And Stem cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
128
|
Phan-Everson T, Etoc F, Li S, Khodursky S, Yoney A, Brivanlou AH, Siggia ED. Differential compartmentalization of BMP4/NOGGIN requires NOGGIN trans-epithelial transport. Dev Cell 2021; 56:1930-1944.e5. [PMID: 34051144 DOI: 10.1016/j.devcel.2021.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/01/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022]
Abstract
Using self-organizing human models of gastrulation, we previously showed that (1) BMP4 initiates the cascade of events leading to gastrulation, (2) BMP4 signal reception is restricted to the basolateral domain, and (3) in a human-specific manner, BMP4 directly induces the expression of NOGGIN. Here, we report the surprising discovery that in human epiblasts, NOGGIN and BMP4 were secreted into opposite extracellular spaces. Interestingly, apically presented NOGGIN could inhibit basally delivered BMP4. Apically imposed microfluidic flow demonstrated that NOGGIN traveled in the apical extracellular space. Our co-localization analysis detailed the endocytotic route that trafficked NOGGIN from the apical space to the basolateral intercellular space where BMP4 receptors were located. This apical-basal transcytosis was indispensable for NOGGIN inhibition. Taken together, the segregation of activator/inhibitor into distinct extracellular spaces challenges classical views of morphogen movement. We propose that the transport of morphogen inhibitors regulates the spatial availability of morphogens during embryogenesis.
Collapse
Affiliation(s)
- Tien Phan-Everson
- Laboratory of Stem Cell Biology and Molecular Embryology, the Rockefeller University, New York, NY 10065, USA; Center for Studies in Physics and Biology, the Rockefeller University, New York, NY 10065, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, the Rockefeller University, New York, NY 10065, USA
| | - Shu Li
- Laboratory of Stem Cell Biology and Molecular Embryology, the Rockefeller University, New York, NY 10065, USA
| | - Samuel Khodursky
- Center for Studies in Physics and Biology, the Rockefeller University, New York, NY 10065, USA
| | - Anna Yoney
- Laboratory of Stem Cell Biology and Molecular Embryology, the Rockefeller University, New York, NY 10065, USA; Center for Studies in Physics and Biology, the Rockefeller University, New York, NY 10065, USA; Department of Genetics and Development, Columbia University, New York, NY 10032
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, the Rockefeller University, New York, NY 10065, USA.
| | - Eric D Siggia
- Center for Studies in Physics and Biology, the Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
129
|
Spiteri C, Caprettini V, Chiappini C. Biomaterials-based approaches to model embryogenesis. Biomater Sci 2021; 8:6992-7013. [PMID: 33136109 DOI: 10.1039/d0bm01485k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding, reproducing, and regulating the cellular and molecular processes underlying human embryogenesis is critical to improve our ability to recapitulate tissues with proper architecture and function, and to address the dysregulation of embryonic programs that underlies birth defects and cancer. The rapid emergence of stem cell technologies is enabling enormous progress in understanding embryogenesis using simple, powerful, and accessible in vitro models. Biomaterials are playing a central role in providing the spatiotemporal organisation of biophysical and biochemical signalling necessary to mimic, regulate and dissect the evolving embryonic niche in vitro. This contribution is rapidly improving our understanding of the mechanisms underlying embryonic patterning, in turn enabling the development of more effective clinical interventions for regenerative medicine and oncology. Here we highlight how key biomaterial approaches contribute to organise signalling in human embryogenesis models, and we summarise the biological insights gained from these contributions. Importantly, we highlight how nanotechnology approaches have remained largely untapped in this space, and we identify their key potential contributions.
Collapse
Affiliation(s)
- Chantelle Spiteri
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| | | | | |
Collapse
|
130
|
Posfai E, Lanner F, Mulas C, Leitch HG. All models are wrong, but some are useful: Establishing standards for stem cell-based embryo models. Stem Cell Reports 2021; 16:1117-1141. [PMID: 33979598 PMCID: PMC8185978 DOI: 10.1016/j.stemcr.2021.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Detailed studies of the embryo allow an increasingly mechanistic understanding of development, which has proved of profound relevance to human disease. The last decade has seen in vitro cultured stem cell-based models of embryo development flourish, which provide an alternative to the embryo for accessible experimentation. However, the usefulness of any stem cell-based embryo model will be determined by how accurately it reflects in vivo embryonic development, and/or the extent to which it facilitates new discoveries. Stringent benchmarking of embryo models is thus an important consideration for this growing field. Here we provide an overview of means to evaluate both the properties of stem cells, the building blocks of most embryo models, as well as the usefulness of current and future in vitro embryo models.
Collapse
Affiliation(s)
- Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| | - Fredrik Lanner
- Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden; Ming Wai Lau Center for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Carla Mulas
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Harry G Leitch
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK; Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London W2 1PG, UK
| |
Collapse
|
131
|
Morales JS, Raspopovic J, Marcon L. From embryos to embryoids: How external signals and self-organization drive embryonic development. Stem Cell Reports 2021; 16:1039-1050. [PMID: 33979592 PMCID: PMC8185431 DOI: 10.1016/j.stemcr.2021.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Embryonic development has been traditionally seen as an inductive process directed by exogenous maternal inputs and extra-embryonic signals. Increasing evidence, however, is showing that, in addition to exogenous signals, the development of the embryo involves endogenous self-organization. Recently, this self-organizing potential has been highlighted by a number of stem cell models known as embryoids that can recapitulate different aspects of embryogenesis in vitro. Here, we review the self-organizing behaviors observed in different embryoid models and seek to reconcile this new evidence with classical knowledge of developmental biology. This analysis leads to reexamine embryonic development as a guided self-organizing process, where patterning and morphogenesis are controlled by a combination of exogenous signals and endogenous self-organization. Finally, we discuss the multidisciplinary approach required to investigate the genetic and cellular basis of self-organization.
Collapse
Affiliation(s)
- J Serrano Morales
- Andalusian Center for Developmental Biology (CABD), CSIC - UPO - JA, Seville, Spain
| | - Jelena Raspopovic
- Andalusian Center for Developmental Biology (CABD), CSIC - UPO - JA, Seville, Spain.
| | - Luciano Marcon
- Andalusian Center for Developmental Biology (CABD), CSIC - UPO - JA, Seville, Spain.
| |
Collapse
|
132
|
Matthews KRW, Wagner DS, Warmflash A. Stem cell-based models of embryos: The need for improved naming conventions. Stem Cell Reports 2021; 16:1014-1020. [PMID: 33770498 PMCID: PMC8185370 DOI: 10.1016/j.stemcr.2021.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/04/2023] Open
Abstract
Stem cell-based models of embryos are known by various names, with different naming conventions, leading to confusion regarding their composition and potential. We propose the need for a general term for the field to promote public engagement and the development of a systematic nomenclature system to differentiate between specific models.
Collapse
Affiliation(s)
- Kirstin R W Matthews
- Baker Institute for Public Policy-Center for Health and Biosciences, Rice University, Houston, TX 77005, USA.
| | - Daniel S Wagner
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Aryeh Warmflash
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| |
Collapse
|
133
|
Minn KT, Dietmann S, Waye SE, Morris SA, Solnica-Krezel L. Gene expression dynamics underlying cell fate emergence in 2D micropatterned human embryonic stem cell gastruloids. Stem Cell Reports 2021; 16:1210-1227. [PMID: 33891870 PMCID: PMC8185470 DOI: 10.1016/j.stemcr.2021.03.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 11/26/2022] Open
Abstract
Human embryonic stem cells cultured in 2D micropatterns with BMP4 differentiate into a radial arrangement of germ layers and extraembryonic cells. Single-cell transcriptomes demonstrate generation of cell types transcriptionally similar to their in vivo counterparts in Carnegie stage 7 human gastrula. Time-course analyses indicate sequential differentiation, where the epiblast arises by 12 h between the prospective ectoderm in the center and the cells initiating differentiation toward extraembryonic fates at the edge. Extraembryonic and mesendoderm precursors arise from the epiblast by 24 h, while nascent mesoderm, endoderm, and primordial germ cell-like cells form by 44 h. Dynamic changes in transcripts encoding signaling components support a BMP, WNT, and Nodal hierarchy underlying germ-layer specification conserved across mammals, and FGF and HIPPO pathways being active throughout differentiation. This work also provides a resource for mining genes and pathways expressed in a stereotyped 2D gastruloid model, common with other species or unique to human gastrulation.
Collapse
Affiliation(s)
- Kyaw Thu Minn
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sabine Dietmann
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Nephrology, Washington University School of Medicine, St. Louis, MO 63110, USA; Institute for Informatics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sarah E Waye
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
134
|
Liu L, Warmflash A. Self-organized signaling in stem cell models of embryos. Stem Cell Reports 2021; 16:1065-1077. [PMID: 33979594 PMCID: PMC8185436 DOI: 10.1016/j.stemcr.2021.03.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Mammalian embryonic development is a complex process driven by self-organization. Understanding how a fertilized egg develops into an embryo composed of more than 200 cell types in precise spatial patterns remains one of the fundamental challenges in biology. Pluripotent stem cells have been used as in vitro models for investigating mammalian development, and represent promising building blocks for regenerative therapies. Recently, sophisticated stem cell-based models that recapitulate early embryonic fate patterning and morphogenesis have been developed. In this article, we review recent advances in stem cell models of embryos in particular focusing on signaling activities underpinning cell fate decisions in space and time.
Collapse
Affiliation(s)
- Lizhong Liu
- Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX 77005, USA; Department of Bioengineering, Rice University, Houston, TX 77005, USA.
| |
Collapse
|
135
|
Shankar V, van Blitterswijk C, Vrij E, Giselbrecht S. From Snapshots to Development: Identifying the Gaps in the Development of Stem Cell-based Embryo Models along the Embryonic Timeline. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004250. [PMID: 33898195 PMCID: PMC8061376 DOI: 10.1002/advs.202004250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/20/2020] [Indexed: 05/05/2023]
Abstract
In recent years, stem cell-based models that reconstruct mouse and human embryogenesis have gained significant traction due to their near-physiological similarity to natural embryos. Embryo models can be generated in large numbers, provide accessibility to a variety of experimental tools such as genetic and chemical manipulation, and confer compatibility with automated readouts, which permits exciting experimental avenues for exploring the genetic and molecular principles of self-organization, development, and disease. However, the current embryo models recapitulate only snapshots within the continuum of embryonic development, allowing the progression of the embryonic tissues along a specific direction. Hence, to fully exploit the potential of stem cell-based embryo models, multiple important gaps in the developmental landscape need to be covered. These include recapitulating the lesser-explored interactions between embryonic and extraembryonic tissues such as the yolk sac, placenta, and the umbilical cord; spatial and temporal organization of tissues; and the anterior patterning of embryonic development. Here, it is detailed how combinations of stem cells and versatile bioengineering technologies can help in addressing these gaps and thereby extend the implications of embryo models in the fields of cell biology, development, and regenerative medicine.
Collapse
Affiliation(s)
- Vinidhra Shankar
- Maastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | | | - Erik Vrij
- Maastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Maastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
136
|
Wang S, Lin CW, Carleton AE, Cortez CL, Johnson C, Taniguchi LE, Sekulovski N, Townshend RF, Basrur V, Nesvizhskii AI, Zou P, Fu J, Gumucio DL, Duncan MC, Taniguchi K. Spatially resolved cell polarity proteomics of a human epiblast model. SCIENCE ADVANCES 2021; 7:7/17/eabd8407. [PMID: 33893097 PMCID: PMC8064645 DOI: 10.1126/sciadv.abd8407] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/05/2021] [Indexed: 05/08/2023]
Abstract
Critical early steps in human embryonic development include polarization of the inner cell mass, followed by formation of an expanded lumen that will become the epiblast cavity. Recently described three-dimensional (3D) human pluripotent stem cell-derived cyst (hPSC-cyst) structures can replicate these processes. To gain mechanistic insights into the poorly understood machinery involved in epiblast cavity formation, we interrogated the proteomes of apical and basolateral membrane territories in 3D human hPSC-cysts. APEX2-based proximity bioinylation, followed by quantitative mass spectrometry, revealed a variety of proteins without previous annotation to specific membrane subdomains. Functional experiments validated the requirement for several apically enriched proteins in cyst morphogenesis. In particular, we found a key role for the AP-1 clathrin adaptor complex in expanding the apical membrane domains during lumen establishment. These findings highlight the robust power of this proximity labeling approach for discovering novel regulators of epithelial morphogenesis in 3D stem cell-based models.
Collapse
Affiliation(s)
- Sicong Wang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amber E Carleton
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chari L Cortez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Linnea E Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nikola Sekulovski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ryan F Townshend
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mara C Duncan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Kenichiro Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
137
|
Depletion of aneuploid cells in human embryos and gastruloids. Nat Cell Biol 2021; 23:314-321. [PMID: 33837289 DOI: 10.1038/s41556-021-00660-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 03/04/2021] [Indexed: 02/01/2023]
Abstract
Chromosomal instability leading to aneuploidy is pervasive in early human embryos1-3 and is considered as a major cause of infertility and pregnancy wastage4,5. Here we provide several lines of evidence that blastocysts containing aneuploid cells are worthy of in vitro fertilization transfer. First, we show clinically that aneuploid embryos can lead to healthy births, suggesting the presence of an in vivo mechanism to eliminate aneuploidy. Second, early development and cell specification modelled in micropatterned human 'gastruloids' grown in confined geometry show that aneuploid cells are depleted from embryonic germ layers, but not from extraembryonic tissue, by apoptosis in a bone morphogenetic protein 4 (BMP4)-dependent manner. Third, a small percentage of euploid cells rescues embryonic tissue in mosaic gastruloids when mixed with aneuploid cells. Finally, single-cell RNA-sequencing analysis of early human embryos revealed a decline of aneuploidy beginning on day 3. Our findings challenge two current dogmas: that a single trophectoderm biopsy at blastocyst stage to perform prenatal genetic testing can accurately determine the chromosomal make-up of a human embryo, and that aneuploid embryos should be withheld from embryo transfer in association with in vitro fertilization.
Collapse
|
138
|
Schwarz C, Hadjantonakis AK. Cells under Tension Drive Gastrulation. Dev Cell 2021; 55:669-670. [PMID: 33352138 DOI: 10.1016/j.devcel.2020.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In this issue of Developmental Cell, Muncie et al. describe the generation of gastrulation-like foci of cells within micropatterned colonies of pluripotent stem cells. This demonstration of mechanosensitive β-catenin/Wnt-dependent specification of cell fate during gastrulation illustrates the insights gleaned by placing stem cells in embryo-like mechanical environments.
Collapse
Affiliation(s)
- Clayton Schwarz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
139
|
Ismagulov G, Hamidi S, Sheng G. Epithelial-Mesenchymal Transition Drives Three-Dimensional Morphogenesis in Mammalian Early Development. Front Cell Dev Biol 2021; 9:639244. [PMID: 33644076 PMCID: PMC7905045 DOI: 10.3389/fcell.2021.639244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/06/2021] [Indexed: 11/20/2022] Open
Abstract
From fertilization to onset of gastrulation, a mammalian embryo goes through several rounds of cellular morphogenesis resembling phenomena of epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET), collectively referred to as EMTs. How these EMT events play a role in shaping the three-dimensional (3-D) architecture of the developing embryo is not well-understood. In this review, we present a model in which cellular morphogenesis, represented primarily by dynamic changes in its epithelialization status, is the driving force of embryonic 3-D organization. This is achieved through the integration of three key components of mammalian early development, the pluripotency regulation, morphogenetic signaling, and biomechanical force anisotropy. Although cells in an early embryo do not exhibit full mesenchymal characteristics, our model underscores the importance of investigating molecular regulation of epithelial cell polarity and partial EMT/MET in understanding mammalian early development.
Collapse
Affiliation(s)
| | | | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
140
|
Zhu M, Cornwall-Scoones J, Wang P, Handford CE, Na J, Thomson M, Zernicka-Goetz M. Developmental clock and mechanism of de novo polarization of the mouse embryo. Science 2021; 370:370/6522/eabd2703. [PMID: 33303584 DOI: 10.1126/science.abd2703] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/14/2020] [Indexed: 12/31/2022]
Abstract
Embryo polarization is critical for mouse development; however, neither the regulatory clock nor the molecular trigger that it activates is known. Here, we show that the embryo polarization clock reflects the onset of zygotic genome activation, and we identify three factors required to trigger polarization. Advancing the timing of transcription factor AP-2 gamma (Tfap2c) and TEA domain transcription factor 4 (Tead4) expression in the presence of activated Ras homolog family member A (RhoA) induces precocious polarization as well as subsequent cell fate specification and morphogenesis. Tfap2c and Tead4 induce expression of actin regulators that control the recruitment of apical proteins on the membrane, whereas RhoA regulates their lateral mobility, allowing the emergence of the apical domain. Thus, Tfap2c, Tead4, and RhoA are regulators for the onset of polarization and cell fate segregation in the mouse.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Jake Cornwall-Scoones
- Division of Biology and Biological Engineering, California Institute of Technology (Caltech), Pasadena, CA 91125, USA
| | - Peizhe Wang
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Charlotte E Handford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Jie Na
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology (Caltech), Pasadena, CA 91125, USA
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK. .,Division of Biology and Biological Engineering, California Institute of Technology (Caltech), Pasadena, CA 91125, USA.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
141
|
Fu J, Warmflash A, Lutolf MP. Stem-cell-based embryo models for fundamental research and translation. NATURE MATERIALS 2021; 20:132-144. [PMID: 33199861 PMCID: PMC7855549 DOI: 10.1038/s41563-020-00829-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/14/2020] [Indexed: 06/01/2023]
Abstract
Despite its importance, understanding the early phases of human development has been limited by availability of human samples. The recent emergence of stem-cell-derived embryo models, a new field aiming to use stem cells to construct in vitro models to recapitulate snapshots of the development of the mammalian conceptus, opens up exciting opportunities to promote fundamental understanding of human development and advance reproductive and regenerative medicine. This Review provides a summary of the current knowledge of early mammalian development, using mouse and human conceptuses as models, and emphasizes their similarities and critical differences. We then highlight existing embryo models that mimic different aspects of mouse and human development. We further discuss bioengineering tools used for controlling multicellular interactions and self-organization critical for the development of these models. We conclude with a discussion of the important next steps and exciting future opportunities of stem-cell-derived embryo models for fundamental discovery and translation.
Collapse
Affiliation(s)
- Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
142
|
Ghimire S, Mantziou V, Moris N, Martinez Arias A. Human gastrulation: The embryo and its models. Dev Biol 2021; 474:100-108. [PMID: 33484705 DOI: 10.1016/j.ydbio.2021.01.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/23/2022]
Abstract
Technical and ethical limitations create a challenge to study early human development, especially following the first 3 weeks of development after fertilization, when the fundamental aspects of the body plan are established through the process called gastrulation. As a consequence, our current understanding of human development is mostly based on the anatomical and histological studies on Carnegie Collection of human embryos, which were carried out more than half a century ago. Due to the 14-day rule on human embryo research, there have been no experimental studies beyond the fourteenth day of human development. Mutagenesis studies on animal models, mostly in mouse, are often extrapolated to human embryos to understand the transcriptional regulation of human development. However, due to the existence of significant differences in their morphological and molecular features as well as the time scale of their development, it is obvious that complete knowledge of human development can be achieved only by studying the human embryo. These studies require a cellular framework. Here we summarize the cellular, molecular, and temporal aspects associated with human gastrulation and discuss how they relate to existing human PSCs based models of early development.
Collapse
Affiliation(s)
- Sabitri Ghimire
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK.
| | - Veronika Mantziou
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Naomi Moris
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | | |
Collapse
|
143
|
Human Embryo Models and Drug Discovery. Int J Mol Sci 2021; 22:ijms22020637. [PMID: 33440617 PMCID: PMC7828037 DOI: 10.3390/ijms22020637] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
For obvious reasons, such as, e.g., ethical concerns or sample accessibility, model systems are of highest importance to study the underlying molecular mechanisms of human maladies with the aim to develop innovative and effective therapeutic strategies. Since many years, animal models and highly proliferative transformed cell lines are successfully used for disease modelling, drug discovery, target validation, and preclinical testing. Still, species-specific differences regarding genetics and physiology and the limited suitability of immortalized cell lines to draw conclusions on normal human cells or specific cell types, are undeniable shortcomings. The progress in human pluripotent stem cell research now allows the growth of a virtually limitless supply of normal and DNA-edited human cells, which can be differentiated into various specific cell types. However, cells in the human body never fulfill their functions in mono-lineage isolation and diseases always develop in complex multicellular ecosystems. The recent advances in stem cell-based 3D organoid technologies allow a more accurate in vitro recapitulation of human pathologies. Embryoids are a specific type of such multicellular structures that do not only mimic a single organ or tissue, but the entire human conceptus or at least relevant components of it. Here we briefly describe the currently existing in vitro human embryo models and discuss their putative future relevance for disease modelling and drug discovery.
Collapse
|
144
|
Muncie JM, Ayad NME, Lakins JN, Xue X, Fu J, Weaver VM. Mechanical Tension Promotes Formation of Gastrulation-like Nodes and Patterns Mesoderm Specification in Human Embryonic Stem Cells. Dev Cell 2020; 55:679-694.e11. [PMID: 33207224 PMCID: PMC7755684 DOI: 10.1016/j.devcel.2020.10.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/20/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022]
Abstract
Embryogenesis is directed by morphogens that induce differentiation within a defined tissue geometry. Tissue organization is mediated by cell-cell and cell-extracellular matrix (ECM) adhesions and is modulated by cell tension and tissue-level forces. Whether cell tension regulates development by modifying morphogen signaling is less clear. Human embryonic stem cells (hESCs) exhibit an intrinsic capacity for self-organization, which motivates their use as a tractable model of early human embryogenesis. We engineered patterned substrates that recapitulate the biophysical properties of the early embryo and mediate the self-organization of "gastrulation-like" nodes in cultured hESCs. Tissue geometries that generated local nodes of high cell-adhesion tension directed the spatial patterning of the BMP4-dependent "gastrulation-like" phenotype by enhancing phosphorylation and junctional release of β-catenin to promote Wnt signaling and mesoderm specification. Furthermore, direct force application via mechanical stretching promoted BMP-dependent mesoderm specification, confirming that tissue-level forces can directly regulate cell fate specification in early human development.
Collapse
Affiliation(s)
- Jonathon M Muncie
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nadia M E Ayad
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Johnathon N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
145
|
Papadopoulos A, Chalmantzi V, Mikhaylichenko O, Hyvönen M, Stellas D, Kanhere A, Heath J, Cunningham DL, Fotsis T, Murphy C. Combined transcriptomic and phosphoproteomic analysis of BMP4 signaling in human embryonic stem cells. Stem Cell Res 2020; 50:102133. [PMID: 33383406 DOI: 10.1016/j.scr.2020.102133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/08/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Human embryonic stem cells (hESCs) are an invaluable tool in the fields of embryology and regenerative medicine. Activin A and BMP4 are well-characterised growth factors implicated in pluripotency and differentiation. In the current study, hESCs are cultured in a modified version of mTeSR1, where low concentrations of ActivinA substitute for TGFβ. This culture system is further used to investigate the changes induced by BMP4 on hESCs by employing a combination of transcriptomic and phosphoproteomic approaches. Results indicate that in a pluripotent state, hESCs maintain WNT signaling under negative regulation by expressing pathway inhibitors. Initial stages of differentiation are characterized by upregulation of WNT pathway ligands, TGFβ pathway inhibitors which have been shown in Xenopus to expand the BMP signaling range essential for embryonic patterning, and mesendodermal transcripts. Moreover, BMP4 enhances the phosphorylation of proteins associated with migration and transcriptional regulation. Results further indicate the vital regulatory role of Activin A and BMP4 in crucial fate decisions in hESCs.
Collapse
Affiliation(s)
- Angelos Papadopoulos
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, United Kingdom
| | - Varvara Chalmantzi
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Olga Mikhaylichenko
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, United Kingdom
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, United Kingdom
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Aditi Kanhere
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - John Heath
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Debbie L Cunningham
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Theodore Fotsis
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, University Campus of Ioannina, 45110 Ioannina, Greece; Laboratory of Biology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Carol Murphy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, University Campus of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
146
|
Tran R, Hoesli CA, Moraes C. Accessible dynamic micropatterns in monolayer cultures via modified desktop xurography. Biofabrication 2020; 13. [PMID: 33238251 DOI: 10.1088/1758-5090/abce0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/25/2020] [Indexed: 11/12/2022]
Abstract
Micropatterned cell cultures provide an important tool to understand dynamic biological processes, but often require specialized equipment and expertise. Here we present subtractive bioscribing (SuBscribe), a readily accessible and inexpensive technique to generate dynamic micropatterns in biomaterial monolayers on-the-fly. We first describe our modifications to a commercially available desktop xurographer and demonstrate the utility and limits of this system in creating micropatterned cultures by mechanically scribing patterns into a brittle, non-adhesive biomaterial layer. Patterns are sufficiently small to influence cell morphology and orientation and can be extended to pattern large areas with complex reproducible shapes. We also demonstrate the use of this system as a dynamic patterning tool for cocultures. Finally, we use this technique to explore and improve upon the well-established epithelial scratch assay, and demonstrate that robotic control of the scratching tool can be used to create custom-shaped wounds in epithelial monolayers, and that the scribing direction leaves trace remnants of matrix molecules that may significantly affect conventional implementations of this common assay.
Collapse
Affiliation(s)
- Raymond Tran
- Department of Chemical Engineering, McGill University, 3610 University Street, Montreal, Quebec, H4X1N3, CANADA
| | - Corinne Annette Hoesli
- Department of Chemical Engineering, McGill University, 3610 University Street, Montreal, Quebec, H4X 1N3, CANADA
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, 3610 University Street, Rm 3A, Montreal, Quebec, H4X1N3, CANADA
| |
Collapse
|
147
|
Minn KT, Fu YC, He S, Dietmann S, George SC, Anastasio MA, Morris SA, Solnica-Krezel L. High-resolution transcriptional and morphogenetic profiling of cells from micropatterned human ESC gastruloid cultures. eLife 2020. [PMID: 33206048 DOI: 10.1101/2020.1101.1122.915777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023] Open
Abstract
During mammalian gastrulation, germ layers arise and are shaped into the body plan while extraembryonic layers sustain the embryo. Human embryonic stem cells, cultured with BMP4 on extracellular matrix micro-discs, reproducibly differentiate into gastruloids, expressing markers of germ layers and extraembryonic cells in radial arrangement. Using single-cell RNA sequencing and cross-species comparisons with mouse, cynomolgus monkey gastrulae, and post-implantation human embryos, we reveal that gastruloids contain cells transcriptionally similar to epiblast, ectoderm, mesoderm, endoderm, primordial germ cells, trophectoderm, and amnion. Upon gastruloid dissociation, single cells reseeded onto micro-discs were motile and aggregated with the same but segregated from distinct cell types. Ectodermal cells segregated from endodermal and extraembryonic but mixed with mesodermal cells. Our work demonstrates that the gastruloid system models primate-specific features of embryogenesis, and that gastruloid cells exhibit evolutionarily conserved sorting behaviors. This work generates a resource for transcriptomes of human extraembryonic and embryonic germ layers differentiated in a stereotyped arrangement.
Collapse
Affiliation(s)
- Kyaw Thu Minn
- Department of Biomedical Engineering, Washington University, St. Louis, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Yuheng C Fu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
- Department of Genetics, Washington University School of Medicine, St. Louis, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, United States
| | - Shenghua He
- Department of Computer Science & Engineering, Washington University, St. Louis, United States
| | - Sabine Dietmann
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
- Division of Nephrology, Washington University School of Medicine, St. Louis, United States
- Institute for Informatics, Washington University School of Medicine, St. Louis, United States
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, United States
| | - Mark A Anastasio
- Department of Biomedical Engineering, Washington University, St. Louis, United States
- Department of Bioengineering, University of Illinois, Urbana-Champaign, United States
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
- Department of Genetics, Washington University School of Medicine, St. Louis, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, United States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
148
|
Minn KT, Fu YC, He S, Dietmann S, George SC, Anastasio MA, Morris SA, Solnica-Krezel L. High-resolution transcriptional and morphogenetic profiling of cells from micropatterned human ESC gastruloid cultures. eLife 2020; 9:e59445. [PMID: 33206048 PMCID: PMC7728446 DOI: 10.7554/elife.59445] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022] Open
Abstract
During mammalian gastrulation, germ layers arise and are shaped into the body plan while extraembryonic layers sustain the embryo. Human embryonic stem cells, cultured with BMP4 on extracellular matrix micro-discs, reproducibly differentiate into gastruloids, expressing markers of germ layers and extraembryonic cells in radial arrangement. Using single-cell RNA sequencing and cross-species comparisons with mouse, cynomolgus monkey gastrulae, and post-implantation human embryos, we reveal that gastruloids contain cells transcriptionally similar to epiblast, ectoderm, mesoderm, endoderm, primordial germ cells, trophectoderm, and amnion. Upon gastruloid dissociation, single cells reseeded onto micro-discs were motile and aggregated with the same but segregated from distinct cell types. Ectodermal cells segregated from endodermal and extraembryonic but mixed with mesodermal cells. Our work demonstrates that the gastruloid system models primate-specific features of embryogenesis, and that gastruloid cells exhibit evolutionarily conserved sorting behaviors. This work generates a resource for transcriptomes of human extraembryonic and embryonic germ layers differentiated in a stereotyped arrangement.
Collapse
Affiliation(s)
- Kyaw Thu Minn
- Department of Biomedical Engineering, Washington UniversitySt. LouisUnited States
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
| | - Yuheng C Fu
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Shenghua He
- Department of Computer Science & Engineering, Washington UniversitySt. LouisUnited States
| | - Sabine Dietmann
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Division of Nephrology, Washington University School of MedicineSt. LouisUnited States
- Institute for Informatics, Washington University School of MedicineSt. LouisUnited States
| | - Steven C George
- Department of Biomedical Engineering, University of California, DavisDavisUnited States
| | - Mark A Anastasio
- Department of Biomedical Engineering, Washington UniversitySt. LouisUnited States
- Department of Bioengineering, University of IllinoisUrbana-ChampaignUnited States
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
149
|
Vermeulen S, Roumans N, Honig F, Carlier A, Hebels DG, Eren AD, Dijke PT, Vasilevich A, de Boer J. Mechanotransduction is a context-dependent activator of TGF-β signaling in mesenchymal stem cells. Biomaterials 2020; 259:120331. [DOI: 10.1016/j.biomaterials.2020.120331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/15/2020] [Accepted: 08/13/2020] [Indexed: 02/08/2023]
|
150
|
Visualization and quantification of dynamic intercellular coupling in human embryonic stem cells using single cell sonoporation. Sci Rep 2020; 10:18253. [PMID: 33106521 PMCID: PMC7589565 DOI: 10.1038/s41598-020-75347-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022] Open
Abstract
Gap junctions (GJs), which are proteinaceous channels, couple adjacent cells by permitting direct exchange of intracellular molecules with low molecular weights. GJ intercellular communication (GJIC) plays a critical role in regulating behaviors of human embryonic stem cells (hESCs), affecting their proliferation and differentiation. Here we report a novel use of sonoporation that enables single cell intracellular dye loading and dynamic visualization/quantification of GJIC in hESC colonies. By applying a short ultrasound pulse to excite single microbubbles tethered to cell membranes, a transient pore on the cell membrane (sonoporation) is generated which allows intracellular loading of dye molecules and influx of Ca2+ into single hESCs. We employ live imaging for continuous visualization of intercellular dye transfer and Ca2+ diffusion in hESC colonies. We quantify cell–cell permeability based on dye diffusion using mass transport models. Our results reveal heterogeneous intercellular connectivity and a variety of spatiotemporal characteristics of intercellular Ca2+ waves in hESC colonies induced by sonoporation of single cells.
Collapse
|